Page 1 of 38

1

Transplantation of heterospheroids of islet cells and mesenchymal stem cells for effective angiogenesis and anti-apoptosis

Jung-Youn Shin1,#, Jee-Heon Jeong2,#, Jin Han1, Suk Ho Bhang3, Gun-Jae Jeong1,

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Muhammad R. Haque4, Taslim A. Al-Hilal5, Myung Kyung Noh1, Youngro Byun4, 5,*, and Byung-Soo Kim1, 6,*

1

School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744,

Republic of Korea. 2

College of Pharmacy, Yeungnam University, Gyeongsan, Gyeongbuk 712-749, Republic of

Korea. 3

School of Chemical Engineering, Sungkyunkwan University, Suwon 440-746, Republic of

Korea. 4

Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of

Convergence Science and Technology, Seoul National University, Seoul 151-742, Republic of Korea. 5

Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National

University, Seoul 151-742, Republic of Korea. 6

Bio-MAX Institute, Institute for Chemical Processes, Engineering Research Institute, Seoul

National University, Seoul 151-744, Republic of Korea.

#

These authors contributed equally to this work.

*

These authors contributed equally as corresponding authors.

1

Page 2 of 38

2

*Author to whom correspondence should be addressed: Youngro Byun, PhD, E-mail: [email protected], Tel: +82-2-880-7866, Fax: +82-2-872-7864. Byung-Soo Kim, PhD, E-mail: [email protected], Tel: +82-2-880-1509, Fax: +82-2-888-

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

1604.

2

Page 3 of 38

3

Abstract Although islet transplantation has been suggested as an alternative therapy for type 1 diabetes, there are efficiency concerns that are attributed to poor engraftment of transplanted islets. Hypoxic condition and delayed vasculogenesis induce necrosis and apoptosis of the

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

transplanted islets. To overcome these limitations in islet transplantation, heterospheroids (HSs), which consist of rat islet cells (ICs) and human bone marrow-derived mesenchymal stem cells (hMSCs) were transplanted to kidney and liver. The HSs cultured under hypoxic condition system exhibited a significant increase in anti-apoptotic gene expression in ICs. hMSCs in the HSs secreted angiogenic and anti-apoptotic proteins. With the HS system, ICs and hMSCs were successfully located in the same area of liver after transplantation of HSs via portal vein, whereas the transplantation of islets and the dissociated hMSCs did not result in localization of transplanted ICs and hMSCs in the same area. HS transplantation resulted in an increase in angiogenesis at the transplantation area and a decrease in the apoptosis of transplanted ICs after transplantation into the kidney subcapsule compared to transplantation of islet cell clusters. Insulin production levels of ICs were higher in the HS transplantation group compared to the ICC transplantation group. The HS system may be a more efficient transplantation method than the conventional methods for the treatment of type 1 diabetes.

Keywords: angiogenesis; heterospheroids; islet cells; islet transplantation; mesenchymal stem cells

3

Page 4 of 38

4

Introduction Islet transplantation is a promising method for the treatment of type 1 diabetes.1 Although the rate of insulin-independence has improved considerably with islet transplantation, the hypoxic condition at the cell transplantation region represents a

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

substantial obstacle to overcome in the early stage of transplantation.2-6 Capillary density of pancreatic islets is approximately 10 times higher than that of the exocrine tissue area in the pancreas for effective insulin secretion response to the glucose level in blood.7,8 However, this specialized vasculature of islets is disrupted as extracellular matrix and vessels are lost, which inhibits the survival of core cells of islets.9 As a result, cell viability and function are compromised.10 The transplanted islets suffer from a hypoxic environment and may lose their viability and function during the early stage of transplantation until vascular network formation occurs approximately 14 days after the transplantation.11 Often, more than 50% of the transplanted islets fail in engraftment and undergo programmed cell death and necrosis caused by hypoxia12; these factors constitute the major causes of islet cell death after transplantation. Mesenchymal stem cells (MSCs) can secrete angiogenic growth factors, thereby contributing to angiogenesis and vasculature stabilization in the cell transplantation region.13,14 Moreover, MSC co-transplantation can help to prevent graft rejection, since MSCs have immune-modulating properties.15,16 Thus, researchers have proposed that isletMSC composite may have beneficial effects toward improving viability of the grafted islet.17,18 Johansson et al. reported that islet composites with endothelial cells and MSCs have beneficial effects on angiogenesis and immune regulation after transplantation.19 Sakata et al. also reported that the co-transplantation of islets with MSCs has the potential benefit of enhancing angiogenesis and improving islet function.20 However, transplanted MSCs would be easily washed out into the bloodstream, especially after transplantation via portal vein of 4

Page 5 of 38

5 the liver, whereas large-sized islets (diameter = 50 - 400 μm) would remain. Thus, locating islet cells (ICs) with MSCs at the transplantations site for their effective interaction would be difficult, and transplantation of a mixture of islets and MSCs would not be an ideal method for producing a positive effect of MSCs on islets. This study describes the transplantation of

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

heterospheroids (HSs), which consist of ICs and MSCs to improve localization of islets with MSCs after transplantation (Fig. 1a), vascularization in the transplantation region, and antiapoptotic activity of the transplanted ICs. To analyze the location of ICs and MSCs, the cells were transplanted via portal vein of the liver. Additionally, to evaluate the vascularization and cell survival, the cells were transplanted into a subcapsule of a kidney rather than a liver because transplanted cells can be generally localized in the injection area in the kidney and easily retrieved for studying angiogenesis and transplanted cell apoptosis in vivo.

Materials and Methods Isolation of pancreatic islets and dissociation of intact islets Pancreatic islets were isolated from Sprague–Dawley (SD) rats (8 weeks of age, male, Orient Bio Inc., Seongnam, Republic of Korea) using the collagenase P (Roche Diagnostics, Mannheim, Germany) digestion method (10 ml of solution (0.8 mg/ml) per pancreas). The islets were cultured in RPMI-1640 medium (Sigma) containing 10% (v/v) fetal bovine serum (FBS, Gibco BRL, Carlsbad, CA), 2 mM sodium bicarbonate, 11 mM glucose (Sigma), 6 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES, Sigma) and 1% (v/v) penicillin/streptomycin (Invitrogen, Carlsbad, CA). Two days after culturing islets, the islets were washed with Hanks' balanced salt solution (HBSS, pH 7.4, Sigma) and dissociated into single cells by suspending them in 0.25% (w/v) trypsin-EDTA (Gibco) at 37°C for 10 min. The dissociated ICs were washed at least twice with cold HBSS. All of the animal experiments were performed with the approval of the Institute of Laboratory Animal 5

Page 6 of 38

6

Resources, Seoul National University (IACUC No. SNU-070822-5).

MSC culture Bone marrow-derived human MSCs (hMSCs, Lonza, Walkersville, MD) were

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

cultured in a 150-mm culture dish with DMEM (Gibco BRL) supplemented with 10% (v/v) FBS (Gibco BRL) and 1% (v/v) penicillin/streptomycin (Gibco BRL). The culture medium was changed every other day.

HS and islet cell cluster (ICC) formation using ICs and hMSCs To generate HSs and ICCs, we used the hanging-drop method with modifications (Fig. 1b).21 HSs were formed at various cell number ratios of ICs to hMSC; 10:1, 2:1 and 1:1. Cell suspension solutions were prepared according to the ratio of the different cells (ICs to hMSCs = 10:1, 2:1 and 1:1), but the total cell number concentration was fixed at 2.7 × 104 cells/ml for each group. ICCs and hMSC spheroids were prepared with 2.7 × 104 ICs/ml and 2.7 × 104 hMSCs/ml, respectively. ICs and hMSCs were suspended in DMEM supplemented with 20% (v/v) FBS and 1% (v/v) penicillin/streptomycin (Gibco BRL). Briefly, 30 μl drops of cell suspension were applied to the inside of the lid of a Petri dish containing phosphate buffered saline (PBS, Sigma) to prevent the cells from drying out. After 72 h of incubation in a 37°C incubator, HSs and ICCs were retrieved using a Pasteur pipette and used for the experiment. The size distributions of HSs and ICCs were determined using Multisizer 3 (Beckman Coulter Inc., Brea, CA) equipped with a 400-μm aperture.

Viability test of HSs and ICCs Live/dead assay (Viability/Cytotoxicity Kit, Molecular Probes, Eugene, OR) was performed for visualizing the viability of HSs and ICCs. HSs and ICCs were stained with 6

Page 7 of 38

7

calcein AM and ethidium homodimer-1 (EthD-1) dye for 15 min in the dark. Calcein AM stains viable cells, which emit strong green fluorescence, and EthD-1 produces red fluorescence when it encounters dead cells. After washes with HBSS, the fluorescence intensity of islets was detected using a fluorescence microscope (Eclipse TE2000-S, Nikon,

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Tokyo, Japan). Quantification of live/dead assays were performed with densitometry using Image J software from National Institutes of Health.

Cell composition analysis of HSs hMSCs were stained with PKH26 (Sigma) prior to the spheroid formation. When the HS formation was completed, HSs were trypsinized and dissociated single cells were directly counted to determine the cell ratio. Then, the nuclei were counterstained with DAPI. The cell ratio was determined by the quantification of cell numbers from a minimum of 15 individual images using a fluorescence microscope (IX71inverted microscope, Olympus, Tokyo, Japan). It is well known that islet cells do not proliferate in vitro22-24 and MSCs rarely proliferate in spheroid due to the contact inhibition 25,26, therefore, the cell ratios of HSs were considered to be constant. To visually analyze the cell distribution in HSs, hMSCs were labeled with PKH26, and ICs were stained with PKH67 (Sigma), prior to HS formation. After the HS formation was completed, HSs were collected and fixed with 4% (w/v) paraformaldehyde (PFA, Sigma) for 30 min. PKH26 and PKH67 labeled cells were visualized using a confocal scanning laser microscope (Carl Zeiss-lSM510, Carl Zeiss International, Oberkochen, Germany). Confocal z-stack images, which were composed of 10 pictures, were merged to produce a picture for the analysis.

Angiogenic factor secretion analysis 7

Page 8 of 38

8

For plating the equal number of ICs, 55 clusters of HSs (10:1)/well and 50 clusters of ICCs/well were plated into a 24-well culture plate containing Millicell culture inserts (Millipore, Billerica, MA). The cells were cultured for 7 days and the cell culture supernate was collected every other day. Angiogenic factors in the supernate were analyzed using

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Proteome Profiler™ Human Angiogenesis Antibody Array (R&D Systems, Minneapolis, MN) according to the manufacturer’s instructions. Cell culture supernate was diluted and mixed with a detection antibody cocktail and incubated with a capture antibody arrayed membrane overnight at 4°C. After washing, the membrane was incubated with HRPconjugated streptavidin and visualized with chemiluminescence (Kodak, Rochester, NY). The level of human vascular endothelial growth factor (VEGF) in the collected cell culture supernate was also determined using an enzyme-linked immunosorbent assay kit (ELISA, R&D Systems) according to the manufacturer’s instructions. In order to ensure that there is no cross-reactivity of the kit to rat VEGF, VEGF release profile of ICCs was analyzed as a control.

Transplantation of HSs and ICCs BALB/C nude mice (Orient Bio Inc.) were anesthetized with an intraperitoneal injection of ketamine (80 mg/kg) and xylazine (16 mg/kg). HSs (10:1, 2,200 clusters) and ICCs (2,000 clusters) were transplanted in the subrenal capsule space (n = 6). To investigate localization of ICs and hMSCs in a liver, 600 clusters of HSs (ICs:hMSCs = 10:1, hMSCs = 4.0×104 cells) or 400 clusters of ICCs with hMSCs (4.0×104 cells) were injected into the portal vein (n = 5). ICs and hMSCs were labeled with PKH26 and PKH67, respectively, prior to transplantation. The surgical procedure was considered successful if no bleeding occurred during the surgical procedure.

8

Page 9 of 38

9

Immunohistochemistry The organs with cell transplantation were retrieved at 14 and 28 days after the treatment. Samples were dehydrated with a graded ethanol series and embedded in paraffin. The specimens were sliced into 4-μm thick sections for histological analysis. For the

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

quantification of microvessel density in the transplantation region, the sections were subjected to immunofluorescent staining with anti-CD31 (Abcam, Cambridge, UK) and antiinsulin (Abcam). After washing with PBS, the slides were incubated with FITC-conjugated (Jackson Immuno Research laboratories, West Grove, PA) and rhodamine-conjugated secondary antibodies (Jackson Immuno Research laboratories) to visualize anti-CD31 and anti-insulin signals, respectively. The sections were mounted using mounting solution containing DAPI (Vecta laboratories, Burlingame, CA). Images were taken with a fluorescence microscope (IX71inverted microscope). Whole mount staining of transplanted islets using anti-CD31 (Abcam) and anti-insulin (Abcam) was performed as described in a previous study.27 The images were taken with a confocal scanning laser microscope (Carl Zeiss-lSM510, Carl Zeiss International).

Histological examination for hMSC localization in liver In order to examine localization of ICs and hMSCs in the liver, the livers were retrieved 7 days after transplantation. Samples were dehydrated with a graded ethanol series and embedded in paraffin. The specimens were sliced into 4-μm thick sections. The sections were mounted using mounting solution containing DAPI (Vecta laboratories) to detect fluorescently labelled hMSCs and ICs. For quantification of hMSC localization with ICs, 200 sections were obtained per liver sample (n = 5 livers per group) and 50 sections were randomly picked and analyzed with a fluorescence microscope (IX71inverted microscope).

9

Page 10 of 38

10

Reverse transcriptase polymerase chain reaction (RT-PCR) Cells or tissue samples were homogenized and lysed in TRIzol reagent (Invitrogen). Total RNA was extracted with chloroform (Sigma) and precipitated with 80% (v/v) isopropanol (Sigma). After the supernatant was removed, the RNA pellet was washed with

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

75% (v/v) ethanol, air-dried and dissolved in 0.1% (v/v) diethyl pyrocarbonate-treated water (Sigma). The total RNA concentration was determined using a NanoDrop spectrometer (ND2000, NanoDrop Technologies, Wilmington, DE). Reverse transcription was performed using 1 μg of pure total RNA and Superscripts II reverse transcriptase (Invitrogen), and the synthesized cDNA was amplified by RT-PCR. Quantitative RT-PCR (qRT-PCR) was performed using the StepOnePlus real-time PCR system (Applied Biosystems, Foster City, CA). TaqMan® Master Mix (Applied Biosystems) was used for the reaction. The profiles of gene expression in ICs of spheroids or transplantation area were quantified with TaqMan Gene Expression Assays (Applied Biosystems). For each target, we used rat glyceraldehyde 3-phosphate

dehydrogenase

(GAPDH):

Rn01775763_g1

(Reference

sequence:

NM_017008.3), Bax: Rn02532082_g1 (Reference sequence: NM_017059.1) and Bcl-2: Rn99999125_m1 (Reference sequence: NM_016993.1). Rat-specific primers were used to investigate the ICs only. The expression level of the target genes was determined using the comparative Ct method, whereby the target was normalized to the endogenous reference (GAPDH).28 Relative gene expressions of ICs in spheroids cultured under normoxic and hypoxic condition were evaluated. All of the data were analyzed using the 2-ΔΔCt method. RTPCR was conducted for 35 cycles of denaturing (94°C, 30 s), annealing (60°C, 45 s), and extension (72°C, 45 s) with a final extension at 72°C for 10 min. PCR products were visualized by electrophoresis on a 1% (w/v) agarose gel using RedSafeTM (iNtRON Biotechnology, Kyungki-Do, Korea) staining. The products were analyzed with a gel documentation system (Gel Logic 2200 Pro, Carestream Health, Rochester, NY), and b-actin 10

Page 11 of 38

11

served as an internal control. The sequences of the primers were as follows: human-specific b-actin: forward primer, 5’-GCA TCT TTC CAG CCT TCC TTC C-3’ and reverse primer, 5’-TCA CCT TCA CCG TTC CAG TTT TT-3’; human-specific hypoxia inducible factor-1α (HIF-1α): forward primer, 5’- ACT TCT GGA TGC TGG TGA TT-3’ and reverse primer,

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

5’- TCC TCG GCT AGT TAG GGT AC-3’. Human-specific primers were used to investigate hMSCs only.

Western blot analysis The samples were lysed using a Dounce homogenizer (Kontes,Vineland, NJ, 50 strokes, 4°C) in ice-cold lysis buffer (15 mM Tris HCl (pH 8.0), 0.25 M sucrose, 15 mM NaCl, 1.5 mM MgCl2, 2.5 mM EDTA, 1 mM EGTA, 1 mM dithiothreitol, 2 mM NaPPi, 1 μg/ml pepstatin A, 2.5 μg/ml aprotinin, 5 μg/ml leupeptin, 0.5 mM phenylmethylsulfonyl fluoride, 0.125 mM Na3VO4, 25 mM NaF, and 10 μM lactacystin). Protein concentration was determined using a bicinchoninic acid (BCA) protein assay (Pierce Biotechnology, Rockford, Il). Equal protein concentrations from each sample were mixed with a sample buffer, loaded, and separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) on a 10% (v/v) resolving gel. The proteins that were separated by SDS-PAGE were transferred to an Immobilon-P membrane (Millipore) and subsequently probed with antibody against caspase-7 (Santa Cruz Biotechnology, Santa Cruz, CA), caspase-3 (LifeSpan Biosciences, Seattle, WA), insulin (Abcam), and β-actin (Abcam) for 1 h at room temperature. Then the membranes were incubated with horseradish peroxidase-conjugated secondary antibody (Santa Cruz) for 1 h at room temperature. The blots were developed using an enhanced chemiluminescence detection system (Amersham Bioscience, Piscataway, NJ). Luminescence was recorded on X-ray film (Fuji super RX, Fujifilm Medical Systems, Tokyo, Japan), and bands were imaged and quantified with an Imaging Densitometer (Bio11

Page 12 of 38

12

Rad, Hercules, CA).

Statistical analysis Quantitative data were expressed as mean ± standard deviation. Statistical analysis

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

comparisons were conducted using Bonferroni test. A value of p < 0.05 was considered to be statistically significant.

Results Formation of ICCs and HSs ICs and hMSCs were aggregated using the hanging-drop method within 3 days. ICCs and HSs showed round-shaped spheroids, and the live/dead assay demonstrated that most of the cells of ICCs and HSs were viable (Fig. 2a). Although a few dead cells were detected in ICCs, almost all of the aggregates emitted strong green fluorescence as an indicator of good viability. The diameters of ICCs, HSs (ICs:hMSCs = 10:1), HSs (2:1) and HSs (1:1) were 81.4 ± 64.3 μm, 78.7 ± 49.7 μm, 100.4 ± 55.7 μm and 80.6 ± 48.3 μm, respectively (Fig 2b). No significant difference among the groups was found. Confocal laser scanning microscopy images showed that HSs were composed of ICs and hMSCs (Fig. 2c). Among the mixture of ICs and hMSCs, the cells that have the same characteristics

preferentially

aggregated

first;

subsequently,

the

cell

aggregates

heterogeneously adhered to each other, forming HSs. A quantitative analysis on the cell composition ratio of ICs to hMSCs showed that HSs preserved the initial ratio of islet cells to hMSCs in cell suspension (Fig. 2d). hMSC ratios to the total cells in HSs (10:1), HSs (2:1), and HSs (1:1) were 14.2 ±9.9%, 33.7 ±8.6%, and 45.9 ±9.3%, respectively.

Attenuated apoptosis of ICs in HSs in vitro 12

Page 13 of 38

13

To evaluate the comparative long-term viability of HSs and ICCs, live/dead analysis was performed on day 7 (Fig. 3a). Cells in spheroids showed comparatively high viability. Cell aggregation did not show much difference in live/dead images among ICCs and HSs. The viability of ICCs and HSs cultured for 2 days under normoxic (20% oxygen) and

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

hypoxic (1% oxygen) conditions was compared (Fig. 3b), and their apoptotic and antiapoptotic gene expression levels were analyzed with qRT-PCR (Fig. 3c). Hypoxic condition was set to mimic the in vivo hypoxic environment after IC transplantation.29,30 The live/dead images of ICCs and HSs under normoxic condition did not show much difference between ICCs and HSs (10:1) (Figs. 3a, b). However, in hypoxic condition, hMSCs in HSs relieved islet apoptosis. The ICC group showed relatively more dead signals compared to the HS group. As shown in figure 3b, under hypoxic condition, the dead signal area of MSC spheroids was significantly smaller that of ICCs. This result suggests that it is highly possible that ICs, rather than MSCs, in HSs show the dead signals in hypoxic condition. The apoptotic and anti-apoptotic gene expressions of rat ICs were investigated using rat specific primers (Fig. 3c). Bax, an apoptotic gene, was downregulated in HSs (10:1), HSs (2:1), and HSs (1:1) compared to ICCs (0.6 ± 0.1, 0.4 ± 0.1, and 0.6 ± 0.1, respectively), whereas Bcl-2, an anti-apoptotic gene, was significantly upregulated in HSs (10:1), HSs (2:1), and HSs (1:1) compared to ICCs (5.3 ± 0.7, 2.0 ± 0.5, and 3.9 ± 0.1, respectively). Spheroids cultured under the hypoxic condition demonstrated that Bcl-2 expression was upregulated in HSs (10:1) compared to ICCs.

Selection of appropriate ratio of ICs to hMSCs in HSs It has been known that cell-cell interaction of ICs is indispensable and is essential for insulin secretory response to control glucose concentration in the blood.31-33 To reinforce the insulin secretion ability of ICs, the HS (10:1) group was selected as the experimental group in 13

Page 14 of 38

14

the following experiments. HSs (10:1) have the least number of hMSCs, which ensure a high extent of islet cell-cell interaction and the highest anti-apoptotic effect on the ICs (Fig. 3).

Angiogenic factor secretion of HSs

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Angiogenic factors produced by HSs (10:1) were investigated with a human angiogenesis antibody array (Fig. 4a). The collected cell culture supernate contained numerous angiogenic factors: tissue inhibitor of metalloproteinase-1 (TIMP-1), insulin-like growth factor-binding protein-3 (IGFBP-3), interleukin-8 (CXCl8) and VEGF. The VEGF secretion profile was also investigated with human VEGF ELISA kit (Fig. 4b). The concentration of VEGF in the HS group increased gradually in normoxic and hypoxic conditions. Relatively higher quantity of VEGF was secreted from MSCs under hypoxic culture condition. hMSCs in HSs expressed higher level of HIF-1α mRNA compared to monolayer MSCs, indicating hypoxic condition in spheroids at day 3 (Fig. 4c).

Localization of ICs and hMSCs in the liver Either the ICCs with dissociated hMSCs or the HSs (10:1) alone were transplanted via portal vein of the liver, and the livers were retrieved 7 days after the transplantation. In average, 10 - 30 clusters were found in each liver section, and the percentage of both PKH67positive (hMSC) and PKH26-positive (IC) clusters were evaluated from total clusters in each liver. The result indicated that intraportally transplanted HSs showed localization of ICs with hMSCs in the same area of the liver (Fig. 5). However, when ICCs and dissociated hMSCs were transplanted, hMSCs were not observed near the transplanted ICCs. This result indicated that only the HS transplantation can systemically localize the hMSCs around the ICs in the transplantation site, and can maximize the hMSC effects on islets at posttransplantation. 14

Page 15 of 38

15

Enhanced angiogenesis near ICs by transplantation of HSs The angiogenic ability of transplanted ICCs and HSs (10:1) was evaluated by determining the density of CD31-positive cells in insulin-positive transplanted islet area on

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

days 14 and 28 of transplantation (Fig. 6a). On day 14, CD31-positive cells were detected in the HS-transplanted kidney group. On day 28, the density of CD31-positive cells was significantly increased in the HS transplantation group compared to the ICC transplantation group (Figs. 6a, b). In the ICC transplantation group, CD31-positive cells were slightly detected in the transplantation area on day 28, but the density of the ICC transplantation group was lower than that of the HS transplantation group. These results demonstrate that localization of ICs with MSCs stimulated the vascularization in the islet transplantation area by the angiogenic growth factors released from hMSCs.

Attenuated apoptosis and higher insulin content of ICs by HS transplantation The rat-specific gene expression of Bcl-2 (Fig. 7a) and protein expression of cleaved caspase-3, caspase-3, caspase-7 (Fig. 7b) and insulin (Fig. 7c) of the kidney tissue in the cell transplantation region harvested on days 3 and 7 after the transplantation were analyzed to evaluate the apoptosis and insulin secretion of transplanted rat ICs. In the HS (10:1) transplantation group, rat Bcl-2 mRNA expression was increased to 1.6 ± 0.8-fold on day 3 and significantly increased to 3.0 ± 0.2-fold on day 7 compared to the ICC transplantation group (Fig. 7a). The protein expression levels of cleaved caspase-3, caspase-3 and caspase-7 were significantly attenuated in the HS (10:1) transplantation group on days 3 and 7 compared to ICCs transplantation group. Moreover, the insulin content of ICs was highly maintained in the HS transplantation group at day 3 and 7 compared to the ICC transplantation group. 15

Page 16 of 38

16

Discussion In the present study, the islet-MSC HS system decreased apoptosis of islets after transplantation (Fig. 7), which could be attributed to factors secreted by MSCs. Previously,

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Jung et al. reported that co-culture of islets and MSCs promoted viability and insulin secretory function of islets.34 Park et al. also showed that MSCs secreted various growth factors and increased the viability and the functionality of islets after transplantation.35 One of the factors secreted by MSCs in HSs, which are responsible for the decreased apoptosis of islets, may be VEGF, a pro-angiogenic and anti-apoptotic factor.36 VEGF secretion from MSCs in HSs could be stimulated by the mild hypoxic condition in spheroids37,38 and the insulin secreted by islet cells in HSs40,41. The upregulation of HIF-1α (Fig. 4c), which can be triggered by the mild hypoxic condition in spheroids, stimulates VEGF expression. Recent studies have shown that HIF-1α can be activated in normoxic conditions in response to various peptide mediators, such as, insulin and insulin-like growth factors. Insulin increases the binding affinity of HIF-1α to hypoxia-response elements of VEGF and stimulates VEGF secretion.39 The transplantation of HSs, which is composed of ICs and MSCs, promoted vascularization near the transplanted ICs (Fig. 6), which may improve the viability of the transplanted ICs. The main cause for engraftment failure in islet transplantation is islet damage from the hypoxic condition at the transplantation site. During the islet isolation procedure, islets lose their vasculature of pancreatic islets. Consequently, islets are susceptible to hypoxic injury in the early periods of transplantation. Generally, blood vessel formation begins 2 to 4 days after the transplantation and is generally completed after 12 to 14 days.40 Delayed and insufficient revascularization would induce necrosis of the transplanted islet cells. Preservation of islet viability is the key to prolonging the survival of 16

Page 17 of 38

17 grafted islets19,41, and early islet vasculature formation is crucial for a better transplantation outcome.42 The HS system enabled a successful localization of ICs with MSCs after transplantation (Fig. 5), which is responsible for the increased vascularization and decreased

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

apoptosis of ICs. When islets and dissociated MSCs were co-transplanted, we found that MSCs were easily washed away from the transplantation site into the bloodstream (Fig. 5), which would not allow for the angiogenic and anti-apoptotic effects of MSCs on the transplanted ICs. In contrast, the HS allowed for the localization of ICs with MSCs after transplantation, and the angiogenic and anti-apoptotic factors secreted by the MSCs in HSs would easily affect the adjacent ICs in HSs. Consequently, HS transplantation showed improved blood vessel network formation in the transplantation area and reduced apoptosis of ICs, which may be responsible for the higher insulin expression in HS transplantation group compared to ICC transplantation group (Figs. 6, 7). In this respect, the localization of ICs with MSCs is significant for a successful engraftment and effective insulin secretion outcome of islet transplantation. Therefore, the HS system may be an advantageous delivery method for producing positive effects of MSCs on islet cells, in contrast to the transplantation of a simple mixture of islets and dissociated MSCs. Further studies on evaluating the clinical feasibility of this method are necessary. HS formation requires labor-intensive process of hanging-drop, which should be simplified for the clinical application. Recently, several types of devices have been designed and are commercially available alternatives to the hanging-drop method for the convenience of spheroid formation. These devices include AggreWell™ (STEMCELL Technologies, Vancouver, BC, Canada) and Perfecta3D® 384-Well Hanging Drop Plates (3D Biomatrix, Ann Arbor, MI). Moreover, the long-term survival and functionality of transplanted islets should be examined with glucose-stimulated insulin secretion tests in vivo prior to the clinical 17

Page 18 of 38

18

application. The HS formation technology could be expandable to other cell-based therapy. For example, heterospheroids of endothelial cells and MSCs could improve angiogenesis compared to single cell-type spheroids. Moreover, the HS system can be applied for gene

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

transfection. Transfection of intact islet is difficult even with viral vector, while dissociated cells are more susceptible for gene transfection. With the HS system, genes can be delivered to nuclei of ICs or MSCs to improve cell functions43,44 before the spheroid formation, which can make highly functionalized cell clusters for treatment of type 1 diabetes.

Conclusion ICs and MSCs localized in the same site after transplantation by transplanting HSs. HSs were formed with simple hanging-drop method, and the ratio of ICs to MSCs can be easily modified. ICs successfully localized together with MSCs in the liver by HS transplantation. Moreover, HS transplantation resulted in a significantly lower apoptotic activity of ICs and a higher angiogenesis near ICs than ICC transplantation. Insulin secretion and deposition were higher in the HS transplantation group compared to the ICC transplantation group. With the HS system, islet transplantation may be more efficient compared to the use of conventional transplantation methods for the treatment of type 1 diabetes.

Acknowledgment This work was supported by the Seoul National University Brain Fusion Program Research Grant.

18

Page 19 of 38

19

References

1. Shapiro, A.M., Ricordi, C., Hering, B.J., Auchincloss, H., Lindblad, R., Robertson, R.P., et al. International trial of the Edmonton protocol for islet transplantation. N Engl J Med

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

355, 1318, 2006. 2.

Jeong, J.H., Hong, S.W., Hong, S., Yook, S., Jung, Y., Park, J.B., et al. Surface camouflage of pancreatic islets using 6-arm-PEG-catechol in combined therapy with tacrolimus and anti-CD154 monoclonal antibody for xenotransplantation. Biomaterials 32, 7961, 2011.

3.

Gibly, R.F., Graham, J.G., Luo, X., Lowe, W.L., Jr., Hering, B.J., and Shea, L.D. Advancing islet transplantation: from engraftment to the immune response. Diabetologia 54, 2494, 2011.

4. Goto, M., Tjernberg, J., Dufrane, D., Elgue, G., Brandhorst, D., Ekdahl, K.N., et al. Dissecting

the

instant

blood-mediated

inflammatory

reaction

in

islet

xenotransplantation. Xenotransplantation 15, 225, 2008. 5. Bennet, W., Groth, C.G., Larsson, R., Nilsson, B., and Korsgren, O. Isolated human islets trigger an instant blood mediated inflammatory reaction: implications for intraportal islet transplantation as a treatment for patients with type 1 diabetes. Ups J Med Sci 105, 125, 2000. 6.

Amiji, M., and Park, K. Surface modification of polymeric biomaterials with poly(ethylene oxide), albumin, and heparin for reduced thrombogenicity. J Biomater Sci Polym Ed 4, 217, 1993.

7. Henderson, J.R., and Moss, M.C. A morphometric study of the endocrine and exocrine capillaries of the pancreas. Q J Exp Physiol 70, 347, 1985. 8. Kuroda, M., Oka, T., Oka, Y., Yamochi, T., Ohtsubo, K., Mori, S., et al. Colocalization of 19

Page 20 of 38

20

vascular endothelial growth factor (vascular permeability factor) and insulin in pancreatic islet cells. J Clin Endocrinol Metab 80, 3196, 1995. 9. Emamaullee, J.A., Rajotte, R.V., Liston, P., Korneluk, R.G., Lakey, J.R., Shapiro, A.M., et al. XIAP overexpression in human islets prevents early posttransplant apoptosis and

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

reduces the islet mass needed to treat diabetes. Diabetes 54, 2541, 2005. 10. Thomas, F.T., Contreras, J.L., Bilbao, G., Ricordi, C., Curiel, D., and Thomas, J.M. Anoikis, extracellular matrix, and apoptosis factors in isolated cell transplantation. Surgery 126, 299, 1999. 11. Menger, M.D., Yamauchi, J., and Vollmar, B. Revascularization and microcirculation of freely grafted islets of Langerhans. World J Surg 25, 509, 2001. 12. Biarnes, M., Montolio, M., Nacher, V., Raurell, M., Soler, J., and Montanya, E. Betacell death and mass in syngeneically transplanted islets exposed to short- and long-term hyperglycemia. Diabetes 51, 66, 2002. 13. Jain, R.K. Molecular regulation of vessel maturation. Nature Med 9, 685, 2003. 14. Yancopoulos, G.D., Davis, S., Gale, N.W., Rudge, J.S., Wiegand, S.J. and Holash, J. Vascular-specific growth factors and blood vessel formation. Nature 407, 242, 2000. 15. Madec, A.M., Mallone, R., Afonso, G., Abou Mrad, E., Mesnier, A., Eljaafari, A., et al. Mesenchymal stem cells protect NOD mice from diabetes by inducing regulatory T cells. Diabetologia 52, 1391, 2009. 16. Abdi, R., Fiorina, P., Adra, C.N., Atkinson, M., and Sayegh, M.H. Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes. Diabetes 57, 1759, 2008. 17.

Duprez, I.R., Johansson, U., Nilsson, B., Korsgren, O., and Magnusson, P.U. Preparatory studies of composite mesenchymal stem cell islets for application in intraportal islet transplantation. Ups J Med Sci 116, 8, 2011. 20

Page 21 of 38

21

18. Rackham, C.L., Chagastelles, P.C., Nardi, N.B., Hauge-Evans, A.C., Jones, P.M., and King, A.J.F. Co-transplantation of mesenchymal stem cells maintains islet organisation and morphology in mice. Diabetologia 54, 1127, 2011. 19. Johansson, U., Olsson, A., Gabrielsson, S., Nilsson, B., and Korsgren, O. Inflammatory

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

mediators expressed in human islets of Langerhans: implications for islet transplantation. Biochem Biophys Res Commun 308, 474, 2003. 20. Sakata, N., Chan, N.K., Chrisler, J., Obenaus, A., and Hathout, E. Bone marrow cell cotransplantation

with

islets

improves

their

vascularization

and

function.

Transplantation 89, 686, 2010. 21. Bhang, S.H., Lee, S., Shin, J.Y., Lee, T.J., and Kim, B.S. Transplantation of cord blood mesenchymal stem cells as spheroids enhances vascularization. Tissue Eng Part A 18, 2138, 2012. 22. Atouf, F., Park, C.H., Pechhold, K., Ta, M., Choi, Y., and Lumelsky, N.L. No evidence for mouse pancreatic beta-cell epithelial-mesenchymal transition in vitro. Diabetes 56, 699, 2007. 23.

Chase, L.G., Ulloa-Montoya, F., Kidder, B.L., and Verfaillie, C.M. Islet-derived fibroblast-like cells are not derived via epithelial-mesenchymal transition from Pdx-1 or insulin-positive cells. Diabetes 56, 3, 2007.

24. Weinberg, N., Ouziel-Yahalom, L., Knoller, S., Efrat, S., and Dor, Y. Lineage tracing evidence for in vitro dedifferentiation but rare proliferation of mouse pancreatic betacells. Diabetes 56, 1299, 2007. 25. Abercrombie, M. Contact inhibition and malignancy. Nature 281, 259, 1979. 26. Pardee, A.B. A Restriction Point for Control of Normal Animal Cell Proliferation. Proc Nat Acad Sci USA 71, 1286, 1974. 27.

Laib, A.M., Bartol, A., Alajati, A., Korff, T., Weber, H., and Augustin, H.G. Spheroid21

Page 22 of 38

22

based human endothelial cell microvessel formation in vivo. Nat Protoc 4, 1202, 2009. 28.

La, W.G., Shin, J.Y., Bhang, S.H., Jin, M., Yoon, H.H., Noh, S.S., et al. Culture on a 3,4-dihydroxy-l-phenylalanine-coated surface promotes the osteogenic differentiation of human mesenchymal stem cells. Tissue Eng Part A 19, 1255, 2013.

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

29. Bhang, S.H., Jung, M.J., Shin, J.Y., La, W.G., Hwang, Y.H., Kim, M.J., et al. Mutual effect of subcutaneously transplanted human adipose-derived stem cells and pancreatic islets within fibrin gel. Biomaterials 34, 7247, 2013. 30.

Lehmann, R., Zuellig, R.A., Kugelmeier, P., Baenninger, P.B., Moritz, W., Perren, A., et al. Superiority of small islets in human islet transplantation. Diabetes 56, 594, 2007.

31. Bosco, D., Orci, L., and Meda, P. Homologous but not heterologous contact increases the insulin secretion of individual pancreatic beta cells. Exp Cell Res 184, 72, 1989. 32. Hauge-Evans, A.C., Squires, P.E., Persaud, S.J., and Jones, P.M. Pancreatic beta-cell-tobeta-cell interactions are required for integrated responses to nutrient stimuli: enhanced Ca2+ and insulin secretory responses of MIN6 pseudoislets. Diabetes 481999. 33.

Halban, P.A., Powers, S.L., George, K.L., and Bonner-Weir, S. Spontaneous reassociation of dispersed adult rat pancreatic islet cells into aggregates with threedimensional architecture typical of native islets. Diabetes 36, 783, 1987.

34. Jung, E.J., Kim, S.C., Wee, Y.M., Kim, Y.H., Choi, M.Y., Jeong, S.H., et al. Bone marrow-derived mesenchymal stromal cells support rat pancreatic islet survival and insulin secretory function in vitro. Cytotherapy 13, 19, 2011. 35.

Park, K.S., Kim, Y.S., Kim, J.H., Choi, B., Kim, S.H., Tan, A.H., et al. Trophic molecules derived from human mesenchymal stem cells enhance survival, function, and angiogenesis of isolated islets after transplantation. Transplantation 89, 509, 2010.

36. Nör, J.E., Christensen, J., Mooney, D.J., and Polverini, P.J. Vascular endothelial growth factor (VEGF)-mediated angiogenesis is associated with enhanced endothelial cell 22

Page 23 of 38

23

survival and induction of Bcl-2 expression. Am J Pathol 154, 375, 1999. 37. Bhang, S.H., Cho, S.W., La, W.G., Lee, T.J., Yang, H.S., Sun, A.Y., et al. Angiogenesis in ischemic tissue produced by spheroid grafting of human adipose-derived stromal cells. Biomaterials 32, 2734, 2011.

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

38. Dai, Y., Xu, M., Wang, Y., Pasha, Z., Li, T., and Ashraf, M. HIF-1alpha induced-VEGF overexpression in bone marrow stem cells protects cardiomyocytes against ischemia. J Mol Cell Cardiol 42, 1036, 2007. 39. Treins, C., Giorgetti-Peraldi, S., Murdaca, J., and Van Obberghen, E. Regulation of vascular endothelial growth factor expression by advanced glycation end products. J Biol Chem 276, 43836, 2001. 40.

Griffith, R.C., Scharp, D.W., Hartman, B.K., Ballinger, W.F., and Lacy, P.E. A morphologic study of intrahepatic portal-vein islet isografts. Diabetes 26, 201, 1977.

41. Davalli, A.M., Ogawa, Y., Ricordi, C., Scharp, D.W., Bonner-Weir, S., and Weir, G.C. A selective decrease in the beta cell mass of human islets transplanted into diabetic nude mice. Transplantation 59, 817, 1995. 42. Menger, M.D., Jager, S., Walter, P., Hammersen, F., and Messmer, K. A novel technique for studies on the microvasculature of transplanted islets of Langerhans in vivo. Int J Microcirc Clin Exp 9, 103, 1990. 43. Yook, S., Jeong, J.H., Jung, Y.S., Hong, S.W., Im, B.H., Seo, J.W., et al. Molecularly engineered islet cell clusters for diabetes mellitus treatment. Cell Transplant 21, 1775, 2012. 44. Hasegawa, K., Wakino, S., Kimoto, M., Minakuchi, H., Fujimura, K., Hosoya, K., et al. The hydrolase DDAH2 enhances pancreatic insulin secretion by transcriptional regulation of secretagogin through a Sirt1-dependent mechanism in mice. FASEB J 27, 2301, 2013. 23

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

Page 24 of 38

24

Figure Legends

24

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 25 of 38

25

25

Page 26 of 38

26

Figure 1. Schematic illustration showing (a) an efficient method of co-transplantation of islets and MSCs for improved angiogenesis and islet cell survival and (b) a protocol for the

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

formation of HSs of islet cells and MSCs. (a) Islet transplantation often results in poor islet survival due to hypoxia in the transplantation region. Co-transplantation with MSCs can induce angiogenesis and improve islet cell survival. Importantly, transplantation of HSs of islet cells and MSCs would be more effective for locating islet cells with MSCs, angiogenesis and islet cell survival than in the transplantation of islets and dissociated MSCs. (b) HS consisting of rat ICs and hMSCs can be prepared using the hanging-drop method.

26

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 27 of 38

27

27

Page 28 of 38

28

Figure 2. Characteristic analysis of ICCs and HSs after the complete aggregation. (a) Light microscopic (left) and live/dead cell (right) images of ICCs and HSs. Green and red signals

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

indicate live and dead cells, respectively. Scale bars = 100 μm. (b) The diameters of ICCs and HSs with various ICs/hMSCs ratios of 10:1, 2:1 and 1:1. There was no statistical significance between any two groups. (c) Confocal laser scanning microscopic images of cell clusters (green: ICs; red: hMSCs). Scale bars = 50 μm. (d) Cell images after dissociation of HSs into ICs and hMSCs (left panel, blue: DAPI, red: hMSCs) and cell composition (right panel). Scale bars = 100 μm.

28

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 29 of 38

29

29

Page 30 of 38

30

Figure 3. In vitro viability and apoptosis of cultured HSs, ICCs, and hMSC spheroids. (a) Long-term in vitro viability of ICCs, HSs, and hMSC spheroids on day 7 as evaluated by

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

live/dead assay. Scale bars = 100 μm. (b) Live/dead images (left panel) and their relative quantification graph (right panel) of ICCs, HSs (10:1) and hMSC spheroids cultured under normoxic and hypoxic conditions for 2 days. Scale bars = 100 μm. *p < 0.05 compared to ICCs. (c) Relative expression of Bax and Bcl-2 of ICs in ICCs or HSs cultured under normoxic or hypoxic condition, as evaluated by qRT-PCR. *p < 0.05 compared to ICCs.

30

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 31 of 38

31

31

Page 32 of 38

32

Figure 4. Analysis of angiogenic factors present in cell culture supernate, and human HIF-1α mRNA expressions in monolayer hMSCs and hMSCs of HSs at day 3. (a) The relative

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

amounts of human angiogenic factors (TIMP-1, IGFBP-3, CXCL8 and VEGF) present in cell culture supernate of HSs (10:1). Cell culture supernate was collected on days 3 and 5 and subjected to a human angiogenesis antibody array. Since the data represent the average values from a single test, which yielded 2 test values for each individual cytokine, statistical significance was not determined. (b) The amounts of hVEGF secreted from rat ICCs and HSs (10:1) containing hMSCs and rat ICs cultured in normoxic condition and HSs (10:1) cultured in hypoxic conditions at various culture time points, as evaluated by ELISA. p < 0.05 between any two groups at all time points. (c) Human HIF-1α mRNA expressions of monolayer hMSCs or HSs at day 3.

32

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 33 of 38

33

33

Page 34 of 38

34

Figure 5. ICs and hMSC localization in portal vein of the liver. Prior to transplantation, ICs and hMSCs were labeled with PKH26 (red) and PKH67 (green), respectively. The livers

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

were harvested 7 days after transplantation. Scale bars = 50 μm. The percentage of both PKH67-positive (hMSC) and PKH26-positive (IC) clusters were evaluated from total clusters found in each liver (lower panel). * p < 0.05 compared to HSs (10:1) group.

34

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 35 of 38

35

35

Page 36 of 38

36

Figure 6. Angiogenesis at the transplantation area in the kidney subcapsule after transplantation of both ICCs and HSs (IC:hMSC = 10:1). (a) CD31-positive (green)

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

microvessels with insulin-positive (red) transplanted islet cells (left panel). The kidneys that contained transplanted islets were harvested 14 and 28 days after the transplantation. Arrows indicate CD31-positive microvessels in the transplantation region. Blue indicates nuclei stained with DAPI. Scale bars = 100 μm. Right panel: the density of CD31-positive microvessels in the insulin-positive region. *p < 0.05 compared to ICCs. (b) Whole mount immunostaining of CD31 (green) and insulin (red) in the transplantation area. The kidneys containing transplanted islets were harvested 28 days after the transplantation. Scale bars = 100 μm.

36

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only. Page 37 of 38

37

37

Page 38 of 38

38

Figure 7. Evaluation of apoptosis and insulin content of ICCs and ICs in HSs in the transplantation area of the kidneys. (a) Relative expression of rat-specific Bcl-2 of ICCs and

Tissue Engineering Part A Downloaded from online.liebertpub.com by Erciyes University on 12/31/14. For personal use only.

ICs in HSs (10:1) as evaluated by qRT-PCR. *p < 0.05 compared to ICCs. (b) Apoptotic protein expressions were evaluated by western blot (upper panel) using antibodies against cleaved caspase-3, caspase-3 and caspase-7, and their protein expression levels were evaluated (lower panel). *p < 0.05 compared to ICCs. (c) Insulin content was evaluated by western blot (left panel) using antibodies against insulin, and their protein expression levels were evaluated (right panel). *p < 0.05 compared to HSs.

38

Transplantation of heterospheroids of islet cells and mesenchymal stem cells for effective angiogenesis and antiapoptosis.

Although islet transplantation has been suggested as an alternative therapy for type 1 diabetes, there are efficiency concerns that are attributed to ...
2MB Sizes 0 Downloads 6 Views